Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
PLoS One ; 14(10): e0223209, 2019.
Article in English | MEDLINE | ID: mdl-31665133

ABSTRACT

Friedreich's ataxia (FA) is a neurodegenerative disease with no approved therapy that is the result of frataxin deficiency. The identification of human FA blood biomarkers related to disease severity and neuro-pathomechanism could support clinical trials of drug efficacy. To try to identify human biomarkers of neuro-pathomechanistic relevance, we compared the overlapping gene expression changes of primary blood and skin cells of FA patients with changes in the Dorsal Root Ganglion (DRG) of the KIKO FA mouse model. As DRG is the primary site of neurodegeneration in FA, our goal was to identify which changes in blood and skin of FA patients provide a 'window' into the FA neuropathomechanism inside the nervous system. In addition, gene expression in frataxin-deficient neuroglial cells and FA mouse hearts were compared for a total of 5 data sets. The overlap of these changes strongly supports mitochondrial changes, apoptosis and alterations of selenium metabolism. Consistent biomarkers were observed, including three genes of mitochondrial stress (MTIF2, ENO2), apoptosis (DDIT3/CHOP), oxidative stress (PREX1), and selenometabolism (SEPW1). These results prompted our investigation of the GPX1 activity as a marker of selenium and oxidative stress, in which we observed a significant change in FA patients. We believe these lead biomarkers that could be assayed in FA patient blood as indicators of disease severity and progression, and also support the involvement of mitochondria, apoptosis and selenium in the neurodegenerative process.


Subject(s)
Biomarkers/blood , Friedreich Ataxia/blood , Ganglia, Spinal/metabolism , Oxidative Stress/genetics , Animals , Antioxidants/metabolism , Apoptosis/genetics , Disease Models, Animal , Eukaryotic Initiation Factors/blood , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Ganglia, Spinal/pathology , Gene Expression Regulation/genetics , Guanine Nucleotide Exchange Factors/blood , Humans , Iron-Binding Proteins/genetics , Mice , Mitochondria/metabolism , Mitochondrial Proteins/blood , Myocardium/metabolism , Selenium/metabolism , Transcription Factor CHOP/blood , Frataxin
2.
Proc Natl Acad Sci U S A ; 116(2): 528-533, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30584092

ABSTRACT

The density-regulated protein (DENR) and the malignant T cell-amplified sequence 1 (MCT-1/MCTS1) oncoprotein support noncanonical translation initiation, promote translation reinitiation on a specific set of mRNAs with short upstream reading frames, and regulate ribosome recycling. DENR and MCT-1 form a heterodimer, which binds to the ribosome. We determined the crystal structure of the heterodimer formed by human MCT-1 and the N-terminal domain of DENR at 2.0-Å resolution. The structure of the heterodimer reveals atomic details of the mechanism of DENR and MCT-1 interaction. Four conserved cysteine residues of DENR (C34, C37, C44, C53) form a classical tetrahedral zinc ion-binding site, which preserves the structure of the DENR's MCT-1-binding interface that is essential for the dimerization. Substitution of all four cysteines by alanine abolished a heterodimer formation. Our findings elucidate further the mechanism of regulation of DENR-MCT-1 activities in unconventional translation initiation, reinitiation, and recycling.


Subject(s)
Cell Cycle Proteins/chemistry , Eukaryotic Initiation Factors/chemistry , Oncogene Proteins/chemistry , Protein Multimerization , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Humans , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Structure, Quaternary
3.
Fish Physiol Biochem ; 43(6): 1657-1675, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28913664

ABSTRACT

This study was conducted to evaluate the effect of enzyme-treated soy protein (ETSP) supplementation in the low-protein diet on growth performance, digestive and absorptive capacities, and related signaling molecules' gene expressions in juvenile Jian carp. The results showed that percent weight gain (PWG), specific growth rate (SGR), and feed intake (FI) were decreased by reducing dietary protein from 34 to 32% (P < 0.05). Supplying low-protein diet with optimal ETSP increased previously mentioned indices of juvenile Jian carp (P < 0.05), which also had no significant difference with the high-protein diet (34%CP) (P > 0.05). Compared with the low-protein diet, appropriate ETSP supplementation in the low-protein diet increased (P < 0.05) (1) the trypsin, lipase, and amylase activities in the hepatopancreas; (2) cholecystokinin concentration in the proximal intestine; (3) the γ-glutamyl transpeptidase (γ-GT), alkaline phosphatase (AKP), and Na+/K+-ATPase activities in all intestinal segments; and (4) the messenger RNA (mRNA) levels of trypsin, lipase, and amylase in hepatopancreas and γ-GT in the mid (MI) and distal (DI) intestine, alkaline phosphatase in MI, and Na+/K+-ATPase and target of rapamycin in all intestinal segments. At the same time, appropriate ETSP supplementation in the low-protein diet downregulated the mRNA levels of AKP in the DI and eIF4E-binding protein 2 in all intestinal segments (P < 0.05). In conclusion, adding 10 g ETSP/kg diet in the low-protein diet can restore the growth performance and digestive and absorptive abilities to the levels in group with 34% dietary protein. Supplementation of optimal ETSP in the low-protein diet enhanced the digestive and absorptive abilities and regulated the signaling molecules related to the TOR signaling pathway.


Subject(s)
Carps/physiology , Dietary Proteins/administration & dosage , Digestion/drug effects , Soybean Proteins/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Diet/veterinary , Dietary Supplements , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation/drug effects , Intestinal Mucosa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics
4.
Amino Acids ; 49(5): 957-964, 2017 05.
Article in English | MEDLINE | ID: mdl-28260165

ABSTRACT

L-Arginine has been reported to enhance brown adipose tissue developments in fetal lambs of obese ewes, but the underlying mechanism is unknown. The present study tested the hypothesis that L-arginine stimulates growth and development of brown adipocyte precursor cells (BAPCs) through activation of mammalian target of rapamycin cell signaling. BAPCs isolated from fetal lambs at day 90 of gestation were incubated   for 6 h in arginine-free DMEM, and then cultured in DMEM with concentrations of 50, 100, 200, 500 or 1000 µmol L-arginine/L for 24-96 h. Cell proliferation, protein turnover, the mammalian target of rapamycin (mTOR) signaling pathway and pre-adipocyte differentiation markers were determined. L-arginine treatment enhanced (P < 0.05) BAPC growth and protein synthesis, while inhibiting proteolysis in a dose-dependent manner. Compared with 50 and 100 µmol/L (the concentrations of arginine in the maternal plasma of obese ewes), 200 µmol L-arginine/L (the concentrations of arginine in the maternal plasma of obese ewes receiving arginine supplementation) increased (P < 0.05) the abundances of phosphorylated mTOR, P70S6K and 4EBP1, as well as the abundances of PGC1α, UCP1, BMP7 and PRDM16. These novel findings indicate that increasing extra-cellular arginine concentration from 50 to 200 µmol/L activates mTOR cell signaling in BAPCs and enhances their growth and development in a dose-dependent manner. Our results provide a mechanism for arginine supplementation to enhance the development of brown adipose tissue in fetal lambs.


Subject(s)
Adipocytes, Brown/drug effects , Arginine/pharmacology , Gene Expression Regulation, Developmental , Obesity/genetics , TOR Serine-Threonine Kinases/genetics , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Female , Fetus , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sheep, Domestic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Zinc Fingers/genetics
5.
Sci Rep ; 7: 39825, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28051178

ABSTRACT

Branched-chain amino acids (BCAAs) are essential amino acids for mammals and play key roles in the regulation of protein metabolism. However, the effect of BCAA deficiency on protein metabolism in skeletal muscle in vivo remains unclear. Here we generated mice with lower BCAA concentrations by specifically accelerating BCAA catabolism in skeletal muscle and heart (BDK-mKO mice). The mice appeared to be healthy without any obvious defects when fed a protein-rich diet; however, bolus ingestion of BCAAs showed that mTORC1 sensitivity in skeletal muscle was enhanced in BDK-mKO mice compared to the corresponding control mice. When these mice were fed a low protein diet, the concentration of myofibrillar protein was significantly decreased (but not soluble protein) and mTORC1 activity was reduced without significant change in autophagy. BCAA supplementation in drinking water attenuated the decreases in myofibrillar protein levels and mTORC1 activity. These results suggest that BCAAs are essential for maintaining myofibrillar proteins during protein undernutrition by keeping mTORC1 activity rather than by inhibiting autophagy and translation. This is the first report to reveal the importance of BCAAs for protein metabolism of skeletal muscle in vivo.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Diet, Protein-Restricted , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Protein Kinases/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Dietary Supplements , Eukaryotic Initiation Factors , Kidney/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Myocardium/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
6.
Nutr Res ; 36(9): 925-936, 2016 09.
Article in English | MEDLINE | ID: mdl-27632912

ABSTRACT

Tissue regeneration using adult stem cells (ASCs) has significant potential as a novel treatment for many degenerative diseases. Previous studies have established that age negatively affects the proliferation status and differentiation potential of ASCs, suggesting a possible limitation in their potential therapeutic use. Therefore, we hypothesized that apple extract might exert beneficial effects on ASCs. The specific objectives were to investigate the proliferative effect of apple ethanol extract on human adipose tissue-derived mesenchymal stem cells (ADSCs) and human cord blood-derived mesenchymal stem cells (CB-MSCs), and identify the possible molecular mechanisms. Apple extract promoted proliferation of ADSCs and CB-MSCs as determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Click-iT 5-ethynyl-2'-deoxyuridine flow cytometry assays. In addition, phosphorylation of p44/42 MAPK (ERK), mammalian target of rapamycin (mTOR), p70 S6 kinase (p70S6K), S6 ribosomal protein (S6RP), eukaryotic initiation factor (eIF) 4B and eIF4E was induced stepwise in ADSCs. Furthermore, apple extract significantly induced the production of vascular endothelial growth factor and interleukin-6 in both ADSCs and CB-MSCs. Similarly, apple extract-induced phosphorylation of the mTOR/p70S6K/S6RP/eIF4B/eIF4E pathway was blocked by pretreatment with PD98059, a specific ERK inhibitor. These results indicate that apple extract-induced proliferation of ADSCs under serum-free conditions is mediated by ERK-dependent cytokine production. Moreover, the beneficial effect of apple extract on proliferation of ASCs may overcome the limitation in therapeutic use of stem cells in tissue regeneration and maintenance of stem cell homeostasis.


Subject(s)
Adult Stem Cells/drug effects , Cell Proliferation/drug effects , Malus , Mesenchymal Stem Cells/drug effects , Plant Extracts/pharmacology , Adipose Tissue , Adult , Adult Stem Cells/physiology , Cell Differentiation , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Eukaryotic Initiation Factors/metabolism , Fetal Blood , Humans , Interleukin-6/metabolism , Mesenchymal Stem Cells/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Regeneration , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Tetrazolium Salts , Thiazoles , Vascular Endothelial Growth Factor A/metabolism
7.
Neuroreport ; 27(15): 1182-9, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27584687

ABSTRACT

Granule cell dispersion (GCD) in the dentate gyrus (DG) of the hippocampus is a morphological alteration characteristic of temporal lobe epilepsy. Recently, we reported that treatment with naringin, a flavonoid found in grapefruit and citrus fruits, reduced spontaneous recurrent seizures by inhibiting kainic acid (KA)-induced GCD and neuronal cell death in mouse hippocampus, suggesting that naringin might have beneficial effects for preventing epileptic events in the adult brain. However, it is still unclear whether the beneficial effects of naringin treatment are mediated by the metabolism of naringin into naringenin in the KA-treated hippocampus. To investigate this possibility, we evaluated whether intraperitoneal injections of naringenin could mimic naringin-induced effects against GCD caused by intrahippocampal KA injections in mice. Our results showed that treatment with naringenin delayed the onset of KA-induced seizures and attenuated KA-induced GCD by inhibiting activation of the mammalian target of rapamycin complex 1 in both neurons and reactive astrocytes in the DG. In addition, its administration attenuated the production of proinflammatory cytokines such as tumor necrosis tumor necrosis factor-α (TNFα) and interleukin-1ß (IL-1ß) from microglial activation in the DG following KA treatment. These results suggest that naringenin may be an active metabolite of naringin and help prevent the progression of epileptic insults in the hippocampus in vivo; therefore, naringenin may be a beneficial metabolite of naringin for the treatment of epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Dentate Gyrus/drug effects , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Flavanones/therapeutic use , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cytokines , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy, Temporal Lobe/chemically induced , Eukaryotic Initiation Factors , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism
8.
Phytomedicine ; 23(2): 174-80, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26926179

ABSTRACT

BACKGROUND: Unfolded protein responses (UPR) determine cell fate and are recognized as anticancer targets. In a previous research, we reported that cryptotanshinone (CPT) exerted cytotoxic effects toward acute lymphoblastic leukemia cells through mitochondria-mediated apoptosis. PURPOSE: In the present study, we further investigated the role of UPR in CPT-induced cytotoxicity on acute lymphoblastic leukemia cells by applying tools of pharmacogenomics and bioinformatics. METHODS: Gene expression profiling was performed by mRNA microarray hybridization. Potential transcription factor binding motifs were identified in the promoter regions of the deregulated genes by Cistrome software. Molecular docking on eIF-4A and PI3K was performed to investigate the inhibitory activity of CPT on translation initiation. RESULTS: CPT regulated genes related to UPR and eIF2 signaling pathways. The DNA-Damage-Inducible Transcript 3 (DDIT3) gene, which is activated as consequence of UPR malfunction during apoptosis, was induced and validated by in vitro experiments. Transcription factor binding motif analysis of the microarrary-retrieved deregulated genes in the promoter region emphasized the relevance of transcription factors, such as ATF2, ATF4 and XBP1, regulating UPR and cell apoptosis. Molecular docking suggested inhibitory effects of CPT by binding to eIF-4A and PI3K providing evidence for a role of CPT's in the disruption of protein synthesis. CONCLUSION: CPT triggered UPR and inhibited protein synthesis via eIF-mediated translation initiation, potentially supporting CPT-induced cytotoxic effects toward acute leukemia cells.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Phenanthrenes/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction/drug effects , Unfolded Protein Response/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Computational Biology , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Humans , Molecular Docking Simulation , Pharmacogenetics , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/metabolism , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism
9.
Sci Rep ; 6: 18800, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728896

ABSTRACT

The eukaryotic translation initiation factor 4E (eIF4E) is considered as a key survival protein involved in cell cycle progression, transformation and apoptosis resistance. Herein, we demonstrate that medicinal plant derivative 3-AWA (from Withaferin A) suppressed the proliferation and metastasis of CaP cells through abrogation of eIF4E activation and expression via c-FLIP dependent mechanism. This translational attenuation prevents the de novo synthesis of major players of metastatic cascades viz. c-FLIP, c-Myc and cyclin D1. Moreover, the suppression of c-FLIP due to inhibition of translation initiation complex by 3-AWA enhanced FAS trafficking, BID and caspase 8 cleavage. Further ectopically restored c-Myc and GFP-HRas mediated activation of eIF4E was reduced by 3-AWA in transformed NIH3T3 cells. Detailed underlying mechanisms revealed that 3-AWA inhibited Ras-Mnk and PI3-AKT-mTOR, two major pathways through which eIF4E converges upon eIF4F hub. In addition to in vitro studies, we confirmed that 3-AWA efficiently suppressed tumor growth and metastasis in different mouse models. Given that 3-AWA inhibits c-FLIP through abrogation of translation initiation by co-targeting mTOR and Mnk-eIF4E, it (3-AWA) can be exploited as a lead pharmacophore for promising anti-cancer therapeutic development.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Ehrlich Tumor/genetics , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Carrier Proteins/metabolism , Caspase 8/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/genetics , Copper-Transporting ATPases , Disease Models, Animal , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factors , Humans , Male , Mice , Neoplasm Metastasis , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis/drug effects , Protein Transport , Withanolides/pharmacology , fas Receptor/metabolism
10.
Amino Acids ; 48(2): 523-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26433892

ABSTRACT

Dietary supplementation with N-acetylcysteine (NAC) has been reported to improve intestinal health and treat gastrointestinal diseases. However, the underlying mechanisms are not fully understood. According to previous reports, NAC was thought to exert its effect through glutathione synthesis. This study tested the hypothesis that NAC enhances enterocyte growth and protein synthesis independently of cellular glutathione synthesis. Intestinal porcine epithelial cells were cultured for 3 days in Dulbecco's modified Eagle medium containing 0 or 100 µM NAC. To determine a possible role for GSH (the reduced form of glutathione) in mediating the effect of NAC on cell growth and protein synthesis, additional experiments were conducted using culture medium containing 100 µM GSH, 100 µM GSH ethyl ester (GSHee), diethylmaleate (a GSH-depletion agent; 10 µM), or a GSH-synthesis inhibitor (buthionine sulfoximine, BSO; 20 µM). NAC increased cell proliferation, GSH concentration, and protein synthesis, while inhibiting proteolysis. GSHee enhanced cell proliferation and GSH concentration without affecting protein synthesis but inhibited proteolysis. Conversely, BSO or diethylmaleate reduced cell proliferation and GSH concentration without affecting protein synthesis, while promoting protein degradation. At the signaling level, NAC augmented the protein abundance of total mTOR, phosphorylated mTOR, and phosphorylated 70S6 kinase as well as mRNA levels for mTOR and p70S6 kinase in IPEC-1 cells. Collectively, these results indicate that NAC upregulates expression of mTOR signaling proteins to stimulate protein synthesis in enterocytes independently of GSH generation. Our findings provide a hitherto unrecognized biochemical mechanism for beneficial effects of NAC in intestinal cells.


Subject(s)
Acetylcysteine/pharmacology , Cell Proliferation/drug effects , Enterocytes/metabolism , Glutathione/metabolism , Protein Biosynthesis/drug effects , Animals , Buthionine Sulfoximine/pharmacology , Cell Line , Cysteine/metabolism , Eukaryotic Initiation Factors/metabolism , Glutathione/analogs & derivatives , Glutathione/pharmacology , Maleates/pharmacology , Real-Time Polymerase Chain Reaction , Sus scrofa , TOR Serine-Threonine Kinases/metabolism
11.
Lipids ; 49(12): 1193-201, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25366515

ABSTRACT

The mammalian target of rapamycin (mTOR) signaling pathway is hyperactive in liver, adipose and skeletal muscle tissues of obese rodents. Alpha-lipoic acid (αLA) has been well accepted as a weight-loss treatment, though there are limited studies on its effect on mTOR signaling in high-fat fed, obese rodents. Therefore, the goal of this study was to determine mTOR signaling and oxidative protein alterations in skeletal muscle of high-fat fed, obese rats after αLA supplementation. Phosphorylation of the mTOR substrate, eukaryotic initiation factor (eIF) 4E-binding protein 1 (4E-BP1) and eIF4B were significantly reduced (p < 0.05) in muscle from αLA supplemented rats. Activation of AMP-activated protein kinase (AMPK), an mTOR inhibitory kinase, was higher (p < 0.05) in the αLA group. Protein expression of markers of oxidative metabolism, acetyl CoA carboxylase (ACC), cytochrome c oxidase IV (COX IV), peroxisome proliferator-activated receptor (PPAR), and PPAR gamma coactivator 1-alpha (PGC-1α) were significantly higher (p < 0.05) after αLA supplementation compared to non-supplemented group. Our findings show that αLA supplementation limits the negative ramifications of consuming a high fat diet on skeletal muscle markers of oxidative metabolism and mTORC1 signaling.


Subject(s)
Diet, High-Fat/adverse effects , Multiprotein Complexes/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , TOR Serine-Threonine Kinases/metabolism , Thioctic Acid/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Carrier Proteins/metabolism , Dietary Supplements , Electron Transport Complex IV/metabolism , Eukaryotic Initiation Factors/metabolism , Intracellular Signaling Peptides and Proteins , Male , Mechanistic Target of Rapamycin Complex 1 , Obesity/diet therapy , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphoproteins/metabolism , Phosphorylation/drug effects , Rats, Zucker , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factors/metabolism
12.
Virology ; 468-470: 81-87, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25151062

ABSTRACT

Tomato spotted wilt virus (TSWV) is a negative-strand RNA virus in the family Bunyaviridae and propagates in both insects and plants. Although TSWV can infect a wide range of plant species, host factors involved in viral RNA synthesis of TSWV in plants have not been characterized. In this report, we demonstrate that the cell-free extract derived from one of the host plants can activate mRNA transcriptional activity of TSWV. Based on activity-guided fractionation of the cell-free extract, we identified eukaryotic elongation factor (eEF) 1A as a possible host factor facilitating TSWV transcription and replication. The RNA synthesis-supporting activity decreased in the presence of an eEF1A inhibitor, suggesting that eEF1A plays an important role in RNA synthesis of TSWV.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation, Viral/physiology , RNA, Viral/biosynthesis , Tospovirus/metabolism , Cell Line , Eukaryotic Initiation Factors/genetics , Plant Extracts/chemistry , Plant Proteins/metabolism , RNA, Messenger/biosynthesis , Tospovirus/genetics
13.
Mol Cell Biochem ; 391(1-2): 37-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532005

ABSTRACT

Muscle mass is determined between protein synthesis and protein degradation. Reduction of muscle mass leads to bedridden condition and attenuation of resistance to diseases. Moreover, bedridden condition leads to additional muscle loss due to disuse muscle atrophy. In our previous study (Sato et al. 2013), we showed that administered lysine (Lys), one of essential amino acid, suppressed protein degradation in skeletal muscle. In this study, we investigated that the mechanism of the suppressive effects of Lys on skeletal muscle proteolysis in C2C12 cell line. C2C12 myotubes were incubated in the serum-free medium containing 10 mM Lys or 20 mM Lys, and myofibrillar protein degradation was determined by the rates of 3-methylhistidine (MeHis) release from the cells. The mammalian target of rapamycin (mTOR) activity from the phosphorylation levels of p70-ribosormal protein S6 kinase 1 and eIF4E-binding protein 1 and the autophagic-lysosomal system activity from the ratio of LC3-II/I in C2C12 myotubes stimulated by 10 mM Lys for 0-3 h were measured. The rates of MeHis release were markedly reduced by addition of Lys. The autophagic-lysosomal system activity was inhibited upon 30 min of Lys supplementation. The activity of mTOR was significantly increased upon 30 min of Lys supplementation. The suppressive effect of Lys on the proteolysis by the autophagic-lysosomal system was maintained partially when mTOR activity was inhibited by 100 nM rapamycin, suggesting that some regulator other than mTOR signaling, for example, Akt, might also suppress the autophagic-lysosomal system. From these results, we suggested that Lys suppressed the activity of the autophagic-lysosomal system in part through activation of mTOR and reduced myofibrillar protein degradation in C2C12 myotubes.


Subject(s)
Autophagy/drug effects , Lysine/pharmacology , Lysosomes/metabolism , Muscle Fibers, Skeletal/metabolism , Proteolysis/drug effects , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Eukaryotic Initiation Factors , Gene Expression Regulation/drug effects , Methylhistidines/pharmacology , Mice , Microtubule-Associated Proteins/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myofibrils/drug effects , Myofibrils/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Time Factors , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Arch Dermatol Res ; 305(8): 747-54, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23912479

ABSTRACT

We investigated the proliferative effect of a Acanthopanax senticosus extract (ASE) on human CD49f(+)/CD29(+) keratinocytes and isolated phloridzin from A. senticosus as an active compound. In addition, the possible mechanisms of action were examined. We found that the ASE and phloridzin-promoted proliferation of CD49f(+)/CD29(+) cells using MTT and Click-iT™ EdU flow cytometry assays. In addition, phosphorylation of the p44/42 MAPK (ERK), mTOR, p70 S6 kinase (p70S6K), S6 ribosomal protein (S6RP), eukaryotic initiation factor 4B (eIF4B), and eIF4E was stepwise induced in CD49f(+)/CD29(+) cells. Furthermore, the ASE and phloridzin significantly induced the production of vascular endothelial growth factor and interleukin-6 in CD49f(+)/CD29(+) cells. Similarly, ASE and phloridzin-induced phosphorylation of the mTOR/p70S6K/S6RP/eIF4B/eIF4E pathway was blocked in response to pretreatment with PD98059, a specific ERK inhibitor. Taken together, these results indicate that ASE and phloridzin-induced proliferation of CD49f(+)/CD29(+) cells under serum-free conditions was mediated by the ERK-dependent mTOR pathway.


Subject(s)
Integrin alpha6/metabolism , Integrin beta1/metabolism , Keratinocytes/metabolism , Phlorhizin/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Proliferation , Cells, Cultured , Eleutherococcus , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factors/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Foreskin/cytology , Humans , Male , Phlorhizin/isolation & purification , Phosphorylation , Plant Extracts/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
15.
Food Chem Toxicol ; 59: 303-10, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23811533

ABSTRACT

Microcystins (MCs) are a family of cyclic heptapeptides that are produced by blooming algae Microcystis. MCs have been implicated in the development of liver cancer, necrosis and even intrahepatic bleeding. Effective prophylactic approaches and complete removal of MCs are urgently needed. Accumulating evidence suggests that microcystin-LR (MC-LR)-induced damage is accompanied by oxidative stress. Supplementation of Se can enhance resistance to oxidative stress. Therefore, in the present study, we investigated the protective effects of κ-Selenocarrageenan (Se-Car), a kind of organic Se compound, in Balb/c mice exposed to MC-LR. Our results proved that Se-Car could significantly ameliorate the hepatic damage induced by MC-LR, including serum markers of liver dysfunction, oxidative damages and histological alterations. Furthermore, Se-Car could significantly alleviate the up-regulation of the molecular targets indicating mitochondrial dysfunction and endoplasmic reticulum stress induced by MC-LR. In conclusion, Se-Car showed clear protection against toxicity induced by MC-LR. Thus, Se-Car could be useful as a new category of anti-MC-LR toxicity reagent.


Subject(s)
Antitoxins/therapeutic use , Bacterial Toxins/antagonists & inhibitors , Carrageenan/therapeutic use , Hepatic Insufficiency/prevention & control , Liver/drug effects , Marine Toxins/antagonists & inhibitors , Microcystins/antagonists & inhibitors , Organoselenium Compounds/therapeutic use , Adaptor Proteins, Signal Transducing , Animals , Bacterial Toxins/toxicity , Biomarkers/blood , Carrier Proteins/agonists , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factors , Hepatic Insufficiency/chemically induced , Hepatic Insufficiency/metabolism , Hepatic Insufficiency/pathology , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Marine Toxins/toxicity , Mice , Mice, Inbred BALB C , Microcystins/toxicity , Microcystis/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Oxidative Stress/drug effects , Phosphoproteins/agonists , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Random Allocation , Signal Transduction/drug effects , Survival Analysis
16.
Neurobiol Dis ; 58: 116-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23742760

ABSTRACT

Cellular mechanisms play a role in conversion of the normal prion protein PrP(C) to the disease-associated protein PrP(Sc). The cells provide not only PrP(C), but also still largely undefined factors required for efficient prion replication. Previously, we have observed that interference with ERK and p38-JNK MAP kinase pathways has opposing effects on the formation of prions indicating that the process is regulated by a balance in intracellualar signaling pathways. In order to obtain a "flow-chart" of such pathways, we here studied the activation of MEK/ERK and mTORC1 downstream targets in relation to PrP(Sc) accumulation in GT1-1 cells infected with the RML or 22L prion strains. We show that inhibition of mTORC1 with rapamycin causes a reduction of PrP(Sc) accumulation at similar low levels as seen when the interaction between the translation initiation factors eIF4E and eIF4G downstream mTORC1 is inhibited using 4EGI-1. No effect is seen following the inhibition of molecules (S6K1 and Mnk1) that links MEK/ERK signaling to mTORC1-mediated control of translation. Instead, stimulation (high [KCl] or [serum]) or inhibition (MEK-inhibitor) of prion formation is associated with increased or decreased phosphorylation of the neuronal transcription factor Elk1, respectively. This study shows that prion formation can be modulated by translational initiating factors, and suggests that MEK/ERK signaling plays a role in the conversion of PrP(C) to PrP(Sc) via an Elk1-mediated transcriptional control. Altogether, our studies indicate that prion protein conversion is under the control of intracellular signals, which hypothetically, under certain conditions may elicit irreversible responses leading to progressive neurodegenerative diseases.


Subject(s)
Carrier Proteins/metabolism , Neurons/metabolism , Phosphoproteins/metabolism , Prions/metabolism , ets-Domain Protein Elk-1/metabolism , Adaptor Proteins, Signal Transducing , Animals , Butadienes/pharmacology , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line, Transformed , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factors , Histones/metabolism , Hypothalamus/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Nitriles/pharmacology , Phosphoproteins/genetics , Potassium Chloride/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transfection , ets-Domain Protein Elk-1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Article in English | MEDLINE | ID: mdl-23643758

ABSTRACT

Intracerebroventricular (ICV) injection of ouabain, a specific Na/K-ATPase inhibitor, induces behavioral changes in rats in a putative animal model of mania. The binding of ouabain to Na/K-ATPase affects signaling molecules in vitro, including ERK1/2 and Akt, which promote protein translation. We have also reported that ERK1/2 and Akt in the brain are involved in the ouabain-induced hyperactivity of rats. In this study, rats were given an ICV injection of ouabain, and then their frontal cortices were examined to determine the effects of ouabain on the mTOR/p70S6K/S6 signaling pathway and protein translation, which are important in modifications of neural circuits and behavior. Rats showed ouabain-induced hyperactivity up to 8h following injection, and increased phosphorylation levels of mTOR, p70S6K, S6, eIF4B, and 4E-BP at 1, 2, 4, and 8h following ouabain injection. Immunohistochemical analyses revealed that increased p-S6 immunoreactivity in the cytoplasm of neurons by ouabain was evident in the prefrontal, cingulate, and orbital cortex. These findings suggested increased translation initiation in response to ouabain. The rate of protein synthesis was measured as the amount of [(3)H]-leucine incorporation in the cell-free extracts of frontal cortical tissues, and showed a significant increase at 8h after ouabain injection. These results suggest that ICV injection of ouabain induced activation of the protein translation initiation pathway regulated by ERK1/2 and Akt, and prolonged hyperactivity in rats. In conclusion, protein translation pathway could play an important role in ouabain-induced hyperactivity in a rodent model of mania.


Subject(s)
Enzyme Inhibitors/pharmacology , Frontal Lobe/drug effects , Ouabain/pharmacology , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Carrier Proteins/metabolism , Enzyme Inhibitors/administration & dosage , Eukaryotic Initiation Factors/metabolism , Frontal Lobe/metabolism , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins , Male , Motor Activity/drug effects , Ouabain/administration & dosage , Phosphoproteins/metabolism , Phosphorylation/drug effects , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosome Subunits, Small/metabolism
18.
Autophagy ; 9(5): 781-3, 2013 May.
Article in English | MEDLINE | ID: mdl-23422216

ABSTRACT

MicroRNAs (miRNAs) form a class of ~21 nucleotide (nt) RNAs that post-transcriptionally repress partially complementary messenger RNAs. miRNA-mediated silencing is critical for control of many key biological processes such as tumorigenesis, neuronal synaptic plasticity and defense against bacteria and viruses. Thus, unsurprisingly, miRNA biogenesis, abundance and action are under refined feedback control that is only beginning to be experimentally uncovered. We recently discovered that DICER1 and EIF2C/AGO are targeted for degradation by autophagy as miRNA-free entities by the selective autophagy receptor CALCOCO2/NDP52 (calcium binding and coiled-coil domain 2/nuclear dot protein, 52 kDa). Strikingly, autophagy establishes a checkpoint for continued loading of miRNA, and this checkpoint is required for maintenance of miRNA abundance and proper miRNA activity. This newfound role for autophagy in miRNA biology suggests that human diseases exhibiting misregulated autophagy may be interdependent with defects in miRNA-mediated regulation of gene networks.


Subject(s)
Autophagy/genetics , Homeostasis/genetics , MicroRNAs/metabolism , Eukaryotic Initiation Factors/metabolism , Humans , MicroRNAs/genetics , Models, Biological , RNA Interference , Ribonuclease III/metabolism , Ubiquitination
19.
Free Radic Biol Med ; 53(5): 1048-60, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22796328

ABSTRACT

Oxidative stress is frequently implicated in the pathology of neurodegenerative disease. The chief source of this stress is mitochondrial respiration, via the passage of reducing equivalents through the respiratory chain resulting in a small but potentially pathological production of superoxide. The superoxide that is produced during normal respiration is primarily detoxified within the mitochondria by superoxide dismutase 2 (Sod2), a key protein for maintaining mitochondrial function. Mitochondria are distributed throughout the soma of neurons, as well as along neuronal processes and at the synaptic terminus. This distribution of potentially independent mitochondria throughout the neuron, at distinct subcellular locations, allows for the possibility of regional subcellular deficits in mitochondrial function. There has been increasing interest in the quantification and characterization of messages and proteins at the synapse, because of its importance in neurodegenerative disease, most notably Alzheimer disease. Here, we report the transcriptomic and proteomic changes that occur in synaptosomes from frontal cortices of Sod2 null mice. Constitutively Sod2 null mice were differentially dosed with the synthetic catalytic antioxidant EUK-189, which can extend the life span of these mice, as well as uncovering or preventing neurodegeneration due to endogenous oxidative stress. This approach facilitated insight into the quantification of trafficked messages and proteins to the synaptosome. We used two complementary methods to investigate the nature of the synaptosome under oxidative stress: either whole-genome gene expression microarrays or mass spectrometry-based proteomics using isobaric tagging for relative and absolute quantitation of proteins. We characterized the relative enrichment of gene ontologies at both gene and protein expression levels that occurs from mitochondrial oxidative stress in the synaptosome, which may lead to new avenues of investigation in understanding the regulation of synaptic function in normal and diseased states. As a result of using these approaches, we report for the first time an activation of the mTOR pathway in synaptosomes isolated from Sod2 null mice, confirmed by an upregulation of the phosphorylation of 4E-BP1.


Subject(s)
Mitochondria/metabolism , Oxidative Stress , Proteomics , Synaptosomes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antioxidants/pharmacology , Carrier Proteins/metabolism , Cell Cycle Proteins , Eukaryotic Initiation Factors , Mice , Mice, Knockout , Mitochondria/drug effects , Oligonucleotide Array Sequence Analysis , Organometallic Compounds/pharmacology , Oxidative Stress/drug effects , Phosphoproteins/metabolism , Phosphorylation , RNA, Messenger/drug effects , RNA, Messenger/genetics , Salicylates/pharmacology , Signal Transduction , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Synaptosomes/drug effects , TOR Serine-Threonine Kinases/metabolism
20.
Mol Nutr Food Res ; 56(5): 741-52, 2012 May.
Article in English | MEDLINE | ID: mdl-22648621

ABSTRACT

SCOPE: Nutritional intervention during muscle wasting aims to attenuate net muscle protein loss. Branched chain amino acids, especially leucine, are able to stimulate the anabolic mammalian target of rapamycin (mTOR) signalling cascade and protein synthesis. It has been suggested that muscle myofibrillar protein expression is more responsive to amino acid supplementation compared to cytoplasmic proteins, although accretion of myofibrillar proteins has not extensively been investigated. We hypothesized that leucine specifically increases myofibrillar protein synthesis in skeletal muscle. METHODS AND RESULTS: This hypothesis was investigated in C2C12 skeletal muscle cells using physiologically relevant culture conditions. Leucine supplementation specifically increased myofibrillar protein accretion, including myosin heavy chain-slow and -fast and myosin light chain 1 and -3 in C2C12 cells. Neither total protein content, nor de novo protein synthesis was affected, despite leucine-induced increased 4E-BP1 and S6K1 phosphorylation. Leucine supplementation did not affect myogenesis, measured by creatine kinase activity and myoblast fusion, either. Remarkably, leucine-induced increased myofibrillar protein accretion was accompanied by elevated MyHC mRNA levels, which involved mTOR-dependent and -independent regulation of MyHC-4 and MyHC-7 gene-expression, respectively. CONCLUSION: This study clearly demonstrates myofibrillar and not generic protein accretion in skeletal muscle following leucine supplementation, and suggests this involves pre-translational control of MyHC expression by leucine.


Subject(s)
Leucine/pharmacology , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cells, Cultured , Creatine Kinase/metabolism , Dietary Supplements , Eukaryotic Initiation Factors , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Muscle Development/drug effects , Muscle, Skeletal/cytology , Myofibrils/drug effects , Myofibrils/metabolism , Myosin Heavy Chains/metabolism , Myosin Light Chains/metabolism , Phosphoproteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL